1
|
Zhan Y, Deng Q, Jia Y, Chen Z, Zhao X, Ling Y, Qiu Y, Wang X, Wang F, He M, Huang W, Shen J, Wen S. Pdia3 deficiency exacerbates intestinal injury by disrupting goblet and Paneth cell function during ischemia/reperfusion. Cell Signal 2025; 130:111682. [PMID: 39988288 DOI: 10.1016/j.cellsig.2025.111682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/27/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Intestinal ischemia/reperfusion (I/R) injury is a severe medical condition associated with high mortality rates due to its disruption of intestinal homeostasis and impairment of mucosal defenses. The intestinal epithelium, particularly goblet and Paneth cells, plays a critical role in maintaining gut barrier integrity. Protein disulfide isomerase A3 (PDIA3) is involved in protein folding within intestinal epithelial cells (IECs) and has been linked to the stress response during I/R injury. This study aims to explore the role of PDIA3 in preserving intestinal integrity and immune function during I/R injury. Our study employed both human and mouse models to investigate PDIA3's expression and function. The correlation between PDIA3 expression and disease severity was analyzed using statistical tests, including Pearson's correlation coefficient. An intestinal I/R model was established in intestinal epithelium-specific conditional knockout mice lacking the Pdia3 gene. Single-cell RNA sequencing, immunohistochemistry, and transcriptomic analysis were used to assess PDIA3 expression in various intestinal cell types and to evaluate its role in epithelial differentiation and immune responses. PDIA3 was found to be highly expressed in healthy IECs, especially in goblet and Paneth cells. Its expression was reduced in patients with mesenteric artery ischemia and Pdia3-deficient mice, leading to severe intestinal damage, including impaired goblet and Paneth cell function, reduced antimicrobial peptide production, and altered gut microbiota. Treatment with recombinant defensin α1, an antimicrobial peptide secreted by Paneth cells, significantly alleviated the adverse effects of Pdia3 deficiency, restoring gut microbiota balance and reducing inflammation in the intestinal I/R injury mice. Taken together, our findings suggest that Pdia3 plays a vital role in maintaining intestinal barrier function and immune defense. Its deficiency exacerbates I/R-induced intestinal damage by impairing epithelial differentiation, mucus production, and antimicrobial peptide secretion. Targeting Pdia3 and associated pathways offers promising therapeutic strategies for mitigating I/R injury and restoring intestinal homeostasis.
Collapse
Affiliation(s)
- Yaqing Zhan
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qiwen Deng
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yifan Jia
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Xu Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yihong Ling
- State Key Laboratory of Oncology in South, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuxin Qiu
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiwen Wang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fan Wang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Muchen He
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenqi Huang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Jiantong Shen
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Shihong Wen
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Department of Anesthesiology, Guangxi Hospital Division of the First Affiliated Hospital of Sun Yat-sen University, Nanning, China.
| |
Collapse
|
2
|
Wang F, Wang X, Wang C, Yan W, Xu J, Song Z, Su M, Zeng J, Han Q, Ruan G, Zhang E, Wang W. Gut microbiota-derived glutathione from metformin treatment alleviates intestinal ferroptosis induced by ischemia/reperfusion. BMC Med 2025; 23:285. [PMID: 40361123 PMCID: PMC12076846 DOI: 10.1186/s12916-025-04119-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 05/07/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Intestinal ischemia/reperfusion injury (IIRI) is a life-threatening condition caused by multiple organ and system failures induced by dysbiosis and gut leakage. Metformin has demonstrated efficacy in protecting against IIRI, although the precise role of the gut microbiota in the underlying mechanism is still ambiguous. METHODS This study examined intestinal barrier function and ferroptosis-related parameters in mice with IIRI following treatment with metformin. Additionally, dirty cages and antibiotics were utilized to investigate the impact of the microbiota on the effects of metformin. The analysis included an assessment of the microbial composition of metformin-treated mice and the biosynthetic activity of specific metabolites. RESULTS Metformin effectively reduced gut leakage induced by IIRI, as evidenced by decreased intestinal permeability and increased Occludin, ZO-1, Claudin-1, and MUC-1 expression. A decrease in the expression of the pro-ferroptotic proteins ACSL4, TFR1, and VDAC2/3 and a decrease in dihydroethidium (DHE) fluorescence, iron, malondialdehyde (MDA), and myeloperoxidase (MPO) were further observed in metformin-treated mice. In contrast, the damage to the GPX4/GSH system caused by IIRI was reversed after metformin treatment, as shown by increases in GPX4, SLC7A11, and GSH. The antiferroptotic effects of metformin were phenocopied by its fecal microbiota but were eliminated by antibiotic intake. 16S rRNA analysis revealed that the metformin-modulated gut microbiota was characterized by increased Lactobacillus murinus, which expressed higher levels of GshF that contributed to the mitigation of IIRI. CONCLUSIONS Murine gut microbiota mediated the anti-ferroptotic effect of metformin on IIRI, and the resulting increase in microbial GSH synthesis could serve as a critical pathway for anti-IIRI.
Collapse
Affiliation(s)
- Fangyan Wang
- Institute of Microbiota and Host Inflammation-Related Diseases, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Xinyu Wang
- Department of Pathology, First Affiliated Hospital of Baotou Medical College of Inner, Mongolia University of Science & Technology, Baotou, China
| | - Chaoyi Wang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wangxin Yan
- Department of Colorectal and Anal Surgery, Wenzhou People's Hospital, Wenzhou, China
| | - Junpeng Xu
- Institute of Microbiota and Host Inflammation-Related Diseases, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Zhengyang Song
- Department of Clinical Laboratory, The First Affiliated Hospital of Henan University of Technology, Luoyang, China
| | - Mingli Su
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingjing Zeng
- Institute of Microbiota and Host Inflammation-Related Diseases, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Qiannian Han
- Institute of Microbiota and Host Inflammation-Related Diseases, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Gaoyi Ruan
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Eryao Zhang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wantie Wang
- Institute of Microbiota and Host Inflammation-Related Diseases, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
3
|
Mao QL, Yu ZH, Nie L, Wang FX, Dong YH, Qi XF. Gastrointestinal injury in cardiopulmonary bypass: current insights and future directions. Front Pharmacol 2025; 16:1542995. [PMID: 40356958 PMCID: PMC12067416 DOI: 10.3389/fphar.2025.1542995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Cardiopulmonary bypass (CPB) is an essential component of cardiac surgery. As CPB technology continues to advance and innovate, it has enabled the expansion of surgical boundaries and the resolution of many previously inoperable challenges. However, the occurrence of various complications during CPB warrants attention, with their prevention and management being paramount. The gastrointestinal tract, directly connected to the external environment, is vulnerable not only to external factors but also to internal changes that may induce damage. Both preclinical and clinical research have demonstrated the incidence of gastrointestinal injuries following CPB, often accompanied by dysbiosis and abnormal metabolic outputs. Currently, interventions addressing gastrointestinal injuries following CPB remain insufficient. Although recent years have not seen notable progress in this field, emerging academic research underscores the essential role of the gut microbiome and its metabolic products in sustaining overall health and internal equilibrium. Notably, their significance as the body's "second genome" is increasingly recognized. Consequently, reevaluating the gastrointestinal damage post-CPB, alongside the associated dysbiosis and metabolic disturbances, is imperative. This reassessment carries substantial theoretical and practical implications for enhancing treatment strategies and bettering patient outcomes after CPB. This review aims to deliver a comprehensive synthesis of the latest preclinical and clinical research on CPB, address current challenges and gaps, and explore potential future research directions.
Collapse
Affiliation(s)
- Qi-Long Mao
- Department of Anesthesiology, Bazhong Central Hospital, Bazhong, Sichuan, China
| | - Zi-Hang Yu
- Department of Anesthesiology, Fushun County People’s Hospital, Zigong, Sichuan, China
| | - Liang Nie
- Department of Anesthesiology, Fushun County People’s Hospital, Zigong, Sichuan, China
| | - Fei-Xiang Wang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu-Hui Dong
- Department of Anesthesiology, Bazhong Central Hospital, Bazhong, Sichuan, China
| | - Xiao-Fei Qi
- Department of Anesthesiology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China
- Department of Anesthesiology, Women and Children’s Medical Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China
| |
Collapse
|
4
|
Lin S, Xiao Y, Li S, Tang L, Shi H, Hong M, Ding L. Thiacloprid exposure disrupts the gut-liver axis and induces liver dysfunction in the Reeves' turtles (Mauremys reevesii). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117710. [PMID: 39799922 DOI: 10.1016/j.ecoenv.2025.117710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
As one of the neonicotinoid insecticides, thiacloprid (THI) is extensively used in agriculture and frequently detected in various aquatic environments, posing a potential threat to aquatic organisms. However, the effects of THI exposure on aquatic turtles remain unknown. In this study, we focused on investigating whether THI has a toxic effect on the gut-liver axis in aquatic turtles. The Reeves' turtles (Mauremys reevesii) were exposed to 0.0178 μM, 6 μM, and 60 μM THI for 5 consecutive weeks. The results revealed that THI altered the composition of intestinal flora, with a decrease in the relative abundance of Romboutsia, and an increase in Clostridium_sensu_stricto_1, Cetobacterium, Enterococcus. This disruption of the intestinal barrier led to an increase in lipopolysaccharide (LPS), THI, and other harmful substances entering the liver. Metabolomic and transcriptomic analyses indicated that metabolic dysregulation and differences in gene expression were concentrated in amino acid metabolism and lipid metabolism, ultimately resulting in severe liver damage and steatosis. Furthermore, elevated levels of liver function indicators, including aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), total bile acid (TBA), and triglyceride (TG), were positively correlated with increased THI concentrations. Our findings demonstrate that THI impairs the intestinal barrier and causes liver dysfunction and damage in turtles, providing new insights into evaluating the toxic effects of thiacloprid on aquatic organisms.
Collapse
Affiliation(s)
- Shuqin Lin
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Yunjuan Xiao
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Siyu Li
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Liyan Tang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Haitao Shi
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Meiling Hong
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China.
| | - Li Ding
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China.
| |
Collapse
|
5
|
Chen SY, Xu H, Qin Y, He TQ, Shi RR, Xing YR, Xu J, Cong RC, Wang MR, Yang JS, Gu JH, He BS. Nicotinamide adenine dinucleotide phosphate alleviates intestinal ischemia/reperfusion injury via Nrf2/HO-1 pathway. Int Immunopharmacol 2024; 143:113478. [PMID: 39471691 DOI: 10.1016/j.intimp.2024.113478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/30/2024] [Accepted: 10/20/2024] [Indexed: 11/01/2024]
Abstract
Intestinal ischemia-reperfusion (I/R) injury is a critical condition in the abdomen that has significant morbidity and fatality rates. Prior studies have noted the defensive role of the coenzymatic antioxidant reduced nicotinamide adenine dinucleotide phosphate (NADPH) in heart and brain I/R damage, yet its impact on intestinal I/R trauma required further exploration. Through the application of an in vitro oxygen-glucose deprivation-reoxygenation model and a mouse model of short-term intestinal I/R, this study clarified the defensive mechanisms of NADPH against intestinal I/R injury. We demonstrated that intraperitoneal NADPH administration markedly reduced interleukin-1β (IL-1β) levels and blocked NLRP3 inflammasome activation, hence reducing inflammation. The antioxidative properties of NADPH were established by the reduction of oxidative stress markers and enhancement of glutathione levels. Importantly, NADPH improved intestinal barrier integrity, indicated by an upregulation of zonula occludens-1 and the promotion of a balanced gut microbiome profile. Furthermore, we identified the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1(HO-1) pathway as a crucial conduit for NADPH's beneficence. When this pathway was inhibited by ML385, the favorable outcomes conferred by NADPH were significantly abrogated. These results demonstrate that NADPH functions as an antioxidative, anti-inflammatory, microbiota-balancing, barrier-strengthening, and anti-inflammatory agent against intestinal I/R damage through activation of the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Su-Ying Chen
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Medical School of Nantong University, Nantong 226001, China; Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China; Department of Ultrasonography, Wuxi City Rehabilitation Hospital, Liangxi District Chinese Medicine Hospital, Wuxi 214000, China
| | - Hui Xu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China.
| | - Yan Qin
- Department of Pharmacy, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China
| | - Tian-Qi He
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China; Department of Pharmacy, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China
| | - Rui-Rui Shi
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China
| | - Yu-Run Xing
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Medical School of Nantong University, Nantong 226001, China; Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China
| | - Jian Xu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China; Department of Pharmacy, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China
| | - Ruo-Chen Cong
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Mei-Rong Wang
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Ju-Shun Yang
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Jin-Hua Gu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China; Department of Pharmacy, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong 226001, China.
| | - Bo-Sheng He
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Medical School of Nantong University, Nantong 226001, China; Translational Medicine Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, China.
| |
Collapse
|
6
|
Wang Q, Yu ZH, Nie L, Wang FX, Mu G, Lu B. Assessing the impact of gut microbiota and metabolic products on acute lung injury following intestinal ischemia-reperfusion injury: harmful or helpful? Front Cell Infect Microbiol 2024; 14:1491639. [PMID: 39687547 PMCID: PMC11647003 DOI: 10.3389/fcimb.2024.1491639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Ischemia-reperfusion injury (IRI) is a common and clinically significant form of tissue damage encountered in medical practice. This pathological process has been thoroughly investigated across a variety of clinical settings, including, but not limited to, sepsis, organ transplantation, shock, myocardial infarction, cerebral ischemia, and stroke. Intestinal IRI, in particular, is increasingly recognized as a significant clinical entity due to marked changes in the gut microbiota and their metabolic products, often described as the body's "second genome." These changes in intestinal IRI lead to profound alterations in the gut microbiota and their metabolic outputs, impacting not only the pathology of intestinal IRI itself but also influencing the function of other organs through various mechanisms. Notable among these are brain, liver, and kidney injuries, with acute lung injury being especially significant. This review seeks to explore in depth the roles and mechanisms of the gut microbiota and their metabolic products in the progression of acute lung injury initiated by intestinal IRI, aiming to provide a theoretical basis and directions for future research into the treatment of related conditions.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Anesthesiology, Zigong Fourth People’s Hospital, Zigong, Sichuan, China
| | - Zi-Hang Yu
- Department of Anesthesiology, Fushun County People’s Hospital, Zigong, Sichuan, China
| | - Liang Nie
- Department of Anesthesiology, Fushun County People’s Hospital, Zigong, Sichuan, China
| | - Fei-Xiang Wang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Guo Mu
- Department of Anesthesiology, Zigong Fourth People’s Hospital, Zigong, Sichuan, China
| | - Bin Lu
- Department of Anesthesiology, Zigong Fourth People’s Hospital, Zigong, Sichuan, China
| |
Collapse
|
7
|
Wen LP, Gao SW, Chen HX, Liu Q, Xiao GZ, Lin HC, He QL. Astragaloside IV Ameliorates Colonic Adenomatous Polyps Development by Orchestrating Gut Bifidobacterium and Serum Metabolome. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1527-1554. [PMID: 39164214 DOI: 10.1142/s0192415x24500605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Astragaloside IV (AS-IV), a natural triterpenoid isolated from Astragalus membranaceus, has been used traditionally in Chinese medicine. Previous studies have highlighted its benefits against carcinoma, but its interaction with the gut microbiota and effects on adenomatous polyps are not well understood. This present study investigates the effects of AS-IV on colonic adenomatous polyp (CAP) development in high-fat-diet (HFD) fed [Formula: see text] mice. [Formula: see text] mice were fed an HFD with or without AS-IV or Naringin for 8 weeks. The study assessed CAP proliferation and employed 16S DNA-sequencing and untargeted metabolomics to explore correlations between microbiome and metabolome in CAP development. AS-IV was more effective than Naringin in reducing CAP development, inhibiting colonic proinflammatory cytokines (IL-1β, IL-6, and TNF-α), tumor associated biomarkers (c-Myc, Cyclin D1), and Wnt/β-catenin pathway proteins (Wnt3a, β-catenin). AS-IV also inhibited the proliferative capabilities of human colon cancer cells (HT29, HCT116, and SW620). Multiomics analysis revealed AS-IV increased the abundance of beneficial genera such as Bifidobacterium pseudolongum and significantly modulated serum levels of certain metabolites including linoleate and 2-trans,6-trans-farnesal, which were significantly correlated with the number of CAP. Finally, the anti-adenoma efficacy of AS-IV alone was significantly suppressed post pseudoaseptic intervention in HFD-fed [Formula: see text] mice but could be reinstated following a combined with Bifidobacterium pseudolongum transplant. AS-IV attenuates CAP development in HFD-fed [Formula: see text] mice by regulating gut microbiota and metabolomics, impacting the Wnt3a/β-catenin signaling pathway. This suggests a potential new strategy for the prevention of colorectal cancer, emphasizing the role of gut microbiota in AS-IV's antitumor effects.
Collapse
Affiliation(s)
- Lu-Ping Wen
- School of Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
- Department of Coloproctology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221005, P. R. China
| | - Shao-Wei Gao
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P. R. China
| | - Hua-Xian Chen
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, P. R. China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, P. R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, P. R. China
| | - Qi Liu
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P. R. China
| | - Guo-Zhong Xiao
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, P. R. China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, P. R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, P. R. China
| | - Hong-Cheng Lin
- Department of General Surgery (Department of Coloproctology), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, P. R. China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, P. R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, P. R. China
| | - Qiu-Lan He
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P. R. China
| |
Collapse
|
8
|
Tang J, Zhao M, Miao X, Chen H, Zhao B, Wang Y, Guo Y, Wang T, Cheng X, Ruan H, Zhang J. Bifidobacterium longum GL001 alleviates rat intestinal ischemia-reperfusion injury by modulating gut microbiota composition and intestinal tissue metabolism. Food Funct 2024; 15:3653-3668. [PMID: 38487897 DOI: 10.1039/d3fo03669c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Intestinal ischemia-reperfusion (IIR) injury leads to inflammation and oxidative stress, resulting in intestinal barrier damage. Probiotics, due to their anti-inflammatory and antioxidant properties, are considered for potential intervention to protect the intestinal barrier during IIR injury. Bifidobacterium longum, a recognized probiotic, has targeted effects on IIR injury, but its mechanisms of action are not yet understood. To investigate the mechanism of Bifidobacterium longum intervention in IIR injury, we conducted a study using a rat IIR injury model. The results showed that Bifidobacterium longum could alleviate inflammation and oxidative stress induced by IIR injury by suppressing the NF-κB inflammatory pathway and activating the Keap1/Nrf2 signaling pathway. Bifidobacterium longum GL001 also increased the abundance of the gut microbiota such as Oscillospira, Ouminococcus, Corynebacterium, Lactobacillus, and Akkermansia, while decreasing the abundance of Allobaculum, [Prevotella], Bacteroidaceae, Bacteroides, Shigella, and Helicobacter. In addition, Bifidobacterium longum GL001 reversed the changes in amino acids and bile acids induced by IIR injury and reduced the levels of DL-cysteine, an oxidative stress marker, in intestinal tissue. Spearman correlation analysis showed that L-cystine was positively correlated with Lactobacillus and negatively correlated with Shigella, while DL-proline was positively correlated with Akkermansia. Moreover, bile acids, cholic acid and lithocholic acid, were negatively correlated with Lactobacillus and positively correlated with Shigella. Therefore, Bifidobacterium longum GL001 may alleviate IIR injury by regulating the gut microbiota to modulate intestinal lipid peroxidation and bile acid metabolism.
Collapse
Affiliation(s)
- Jilang Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Mingchao Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Xue Miao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Hong Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Binger Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Yingying Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Yingchao Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Tiantian Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Xin Cheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Hongri Ruan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| | - Jiantao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, China
| |
Collapse
|
9
|
Dicu-Andreescu I, Penescu MN, Verzan C. Septic acute kidney injury and gut microbiome: Should we change our approach? Nefrologia 2024; 44:119-128. [PMID: 38697693 DOI: 10.1016/j.nefroe.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 05/23/2023] [Indexed: 05/05/2024] Open
Abstract
Incidence of acute kidney injury (AKI) remained relatively stable over the last decade and the adjusted risks for it and mortality are similar across different continents and regions. Also, the mortality of septic-AKI can reach 70% in critically-ill patients. These sole facts can give rise to a question: is there something we do not understand yet? Currently, there are no specific therapies for septic AKI and the treatment aims only to maintain the mean arterial pressure over 65mmHg by ensuring a good fluid resuscitation and by using vasopressors, along with antibiotics. On the other hand, there is an increased concern about the different hemodynamic changes in septic AKI versus other forms and the link between the gut microbiome and the severity of septic AKI. Fortunately, progress has been made in the form of administration of pre- and probiotics, short chain fatty acids (SCFA), especially acetate, and also broad-spectrum antibiotics or selective decontaminants of the digestive tract in a successful attempt to modulate the microbial flora and to decrease both the severity of AKI and mortality. In conclusion, septic-AKI is a severe form of kidney injury, with particular hemodynamic changes and with a strong link between the kidney and the gut microbiome. By modulating the immune response we could not only treat but also prevent severe forms. The most difficult part is to categorize patients and to better understand the key mechanisms of inflammation and cellular adaptation to the injury, as these mechanisms can serve in the future as target therapies.
Collapse
Affiliation(s)
- Ioana Dicu-Andreescu
- "Carol Davila" University of Medicine and Pharmacy, str. Eroii Sanitari no. 8, Sector 5, Bucharest, Romania.
| | - Mircea Niculae Penescu
- "Carol Davila" University of Medicine and Pharmacy, str. Eroii Sanitari no. 8, Sector 5, Bucharest, Romania; "Dr. Carol Davila" Clinical Hospital of Nephrology, str. Grivița no. 4, Sector 1, Bucharest, Romania
| | - Constantin Verzan
- "Carol Davila" University of Medicine and Pharmacy, str. Eroii Sanitari no. 8, Sector 5, Bucharest, Romania; "Dr. Carol Davila" Clinical Hospital of Nephrology, str. Grivița no. 4, Sector 1, Bucharest, Romania
| |
Collapse
|
10
|
Zhao M, Tang F, Huang X, Ma J, Wang F, Zhang P. Polysaccharide Isolated from Agaricus blazei Murill Alleviates Intestinal Ischemia/Reperfusion Injury through Regulating Gut Microbiota and Mitigating Inflammation in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:2202-2213. [PMID: 38247134 DOI: 10.1021/acs.jafc.3c08482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Intestinal ischemia-reperfusion (I/R) injury is a serious disease in medical settings, and gut dysbiosis is a major contributor to its development. Polysaccharides from Agaricus blazei Murill (ABM) showed a range of pharmacological activities, yet no studies assessed the potential of ABM polysaccharides for alleviating intestinal I/R injury. Here, we purified a major polysaccharide (ABP1) from an ABM fruit body and subsequently tested its potential to mitigate intestinal I/R injury in a mouse model of temporary superior mesenteric artery occlusion. The results reveal that ABP1 pretreatment enhances gut barrier function via upregulation of the expression of tight junction proteins such as ZO-1 and occludin. Additionally, ABP1 intervention reduces the recruitment of neutrophils and the polarization of M1 macrophages and limits inflammation by blocking the assembly of the NLRP3 inflammasome. Moreover, the role of ABP1 in regulating the gut microbiota was confirmed via antibiotic treatment. The omics data reveals that ABP1 reprograms gut microbiota compositions, characterized by a decrease of Proteobacteria and an increase of Lachnospiraceae and Lactobacillaceae, especially the SCFA-producing genera such as Ligilactobacillus and Blautia. Overall, this work highlights the therapeutic potential of ABP1 against intestinal I/R injury, which mainly exhibits its effects via regulating the gut microbiota and suppressing the overactivated inflammation response.
Collapse
Affiliation(s)
- Meiqi Zhao
- Life and Health Intelligent Research Institute, Tianjin University of Technology, Tianjin 300384, China
- Department of Gastroenterology and Hepatology, Nankai University Affiliated Third Central Hospital, Tianjin 300170, China
| | - Fei Tang
- Department of Gastroenterology and Hepatology, Nankai University Affiliated Third Central Hospital, Tianjin 300170, China
| | - Xiaoyu Huang
- Department of Gastroenterology and Hepatology, Nankai University Affiliated Third Central Hospital, Tianjin 300170, China
| | - Jiajia Ma
- Department of Gastroenterology and Hepatology, Nankai University Affiliated Third Central Hospital, Tianjin 300170, China
| | - Fengmei Wang
- Department of Organ Transplantation, Tianjin Key Laboratory of Organ Transplantation, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
- Department of Gastroenterology and Hepatology, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Peng Zhang
- Life and Health Intelligent Research Institute, Tianjin University of Technology, Tianjin 300384, China
| |
Collapse
|
11
|
Luqman A, Hassan A, Ullah M, Naseem S, Ullah M, Zhang L, Din AU, Ullah K, Ahmad W, Wang G. Role of the intestinal microbiome and its therapeutic intervention in cardiovascular disorder. Front Immunol 2024; 15:1321395. [PMID: 38343539 PMCID: PMC10853344 DOI: 10.3389/fimmu.2024.1321395] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
The gut microbiome is a heterogeneous population of microbes comprising viruses, bacteria, fungi, and protozoa. Such a microbiome is essential for sustaining host equilibrium, and its impact on human health can be altered by a variety of factors such as external variables, social behavior, age, nutrition, and genetics. Gut microbes' imbalances are related to a variety of chronic diseases including cancer, obesity, and digestive disorders. Globally, recent findings show that intestinal microbes have a significant role in the formation of cardiovascular disease (CVD), which is still the primary cause of fatalities. Atherosclerosis, hypertension, diabetes, inflammation, and some inherited variables are all cardiovascular risk variables. However, studies found correlations between metabolism, intestinal flora, and dietary intake. Variations in the diversity of gut microbes and changes in their activity are thought to influence CVD etiology. Furthermore, the gut microbiota acts as an endocrine organ, producing bioactive metabolites such as TMA (trimethylamine)/TMAO (trimethylamine N-oxide), SCFA (short-chain fatty acids), and bile acids, which have a substantial impact on host wellness and disease by multiple mechanisms. The purpose of this overview is to compile current evidence highlighting the intricate links between gut microbiota, metabolites, and the development of CVD. It focuses on how intestinal dysbiosis promotes CVD risk factors such as heart failure, hypertension, and atherosclerosis. This review explores the normal physiology of intestinal microbes and potential techniques for targeting gut bacteria for CVD treatment using various microbial metabolites. It also examines the significance of gut bacteria in disease treatment, including supplements, prebiotics, probiotics, antibiotic therapies, and fecal transplantation, which is an innovative approach to the management of CVD. As a result, gut bacteria and metabolic pathways become increasingly attractive as potential targets for CVD intervention.
Collapse
Affiliation(s)
- Ameer Luqman
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratories, Chongqing, China
| | - Adil Hassan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratories, Chongqing, China
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing University of Science and Technology, Chongqing, China
| | - Mehtab Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Sahar Naseem
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Mehraj Ullah
- School of Fermentation Engineering Tianjin University of Science and Technology, Tianjin, China
| | | | - Ahmad Ud Din
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, United States
| | - Kamran Ullah
- Department of Biology, The University of Haripur, Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Waqar Ahmad
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratories, Chongqing, China
| |
Collapse
|
12
|
Romano A, Balestrini L, Della Scala G, Chico L, Bertini A, Cangioli M, Ciapparelli F, Ciardella E, Congestrí C, Dinelli V, Donati F, Ficini M, Giovanetti C, Nannicini F, Pasquali A, Pieraccini A. Effects of a Nutraceutical Multicompound, with Probiotics, Hericium, PEA, and Undaria in Patients with Irritable Bowel Syndrome. J Diet Suppl 2024; 21:451-461. [PMID: 38186311 DOI: 10.1080/19390211.2023.2296106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal condition. Probiotics and other nutraceutical compounds can have specific indications in the context of IBS. A retrospective analysis was conducted on 123 IBS patients in order to evaluate the effects of an oral probiotic-based dietary supplement (Colicron, one cps/day for 4 wk) on stool consistency and pain intensity. Different time points were defined as follows: baseline (T0), 2 wk of treatment (T2), and 4 wk of treatment (T4). Stool consistency was assessed by using the Bristol Stool Scale. Pain intensity was evaluated by the Visual Analogue Scale (VAS). Patients who were initially categorized as normal retained regular bowel movements throughout the study. Both patients with constipation and diarrhea showed an improvement in the Bristol Stool Scale. The score increased from 1.5 ± 0.5 to 3.3 ± 0.7 (p < 0.001) and decreased from 6.5 ± 0.7 to 4.3 ± 0.9 (p < 0.001) at T4, respectively, compared to T0. The VAS score for pain in the pooled IBS patients improved from 6.7 ± 2.2 to 2.8 ± 1.9 at T0 vs T4 (p < 0.001), with a similar trend also observed when patients were categorized based on stool consistency: normal (from 5.2 ± 1.9 to 2.9 ± 1.7), constipation (from 7.5 ± 1.3 to 3.2 ± 2.2), and diarrhea (6.7 ± 2.3 to 2.5 ± 1.9) (p < 0.001). Colicron could be useful in symptom relief, reducing abdominal pain and improving stool consistency of IBS patients. However, further controlled clinical trials are needed to confirm these preliminary findings.
Collapse
Affiliation(s)
- Antonio Romano
- Gastroenterology Private Practitioner, GF. Medical Center, Pisa, Italy
| | | | | | - Lucia Chico
- Laboratori Aliveda srl, Crespina Lorenzana, Pisa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Modrego J, Ortega-Hernández A, Goirigolzarri J, Restrepo-Córdoba MA, Bäuerl C, Cortés-Macías E, Sánchez-González S, Esteban-Fernández A, Pérez-Villacastín J, Collado MC, Gómez-Garre D. Gut Microbiota and Derived Short-Chain Fatty Acids Are Linked to Evolution of Heart Failure Patients. Int J Mol Sci 2023; 24:13892. [PMID: 37762194 PMCID: PMC10530267 DOI: 10.3390/ijms241813892] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
There is a lack of direct evidence regarding gut microbiota dysbiosis and changes in short-chain fatty acids (SCFAs) in heart failure (HF) patients. We sought to assess any association between gut microbiota composition, SCFA production, clinical parameters, and the inflammatory profile in a cohort of newly diagnosed HF patients. In this longitudinal prospective study, we enrolled eighteen newly diagnosed HF patients. At admission and after 12 months, blood samples were collected for the assessment of proinflammatory cytokines, monocyte populations, and endothelial dysfunction, and stool samples were collected for analysis of gut microbiota composition and quantification of SCFAs. Twelve months after the initial HF episode, patients demonstrated improved clinical parameters and reduced inflammatory state and endothelial dysfunction. This favorable evolution was associated with a reversal of microbiota dysbiosis, consisting of the increment of health-related bacteria, such as genus Bifidobacterium, and levels of SCFAs, mainly butyrate. Furthermore, there was a decrease in the abundance of pathogenic bacteria. In vitro, fecal samples collected after 12 months of follow-up exhibited lower inflammation than samples collected at admission. In conclusion, the favorable progression of HF patients after the initial episode was linked to the reversal of gut microbiota dysbiosis and increased SCFA production, particularly butyrate. Whether restoring butyrate levels or promoting the growth of butyrate-producing bacteria could serve as a complementary treatment for these patients deserves further studies.
Collapse
Affiliation(s)
- Javier Modrego
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.M.); (A.O.-H.); (S.S.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Adriana Ortega-Hernández
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.M.); (A.O.-H.); (S.S.-G.)
| | - Josebe Goirigolzarri
- Servicio de Cardiología, Hospital Clínico de San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.G.); (M.A.R.-C.)
| | - María Alejandra Restrepo-Córdoba
- Servicio de Cardiología, Hospital Clínico de San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.G.); (M.A.R.-C.)
| | - Christine Bäuerl
- Instituto de Agroquímica y Tecnología de los Alimentos (IATA-CSIC), 46980 Paterna, Spain; (C.B.); (E.C.-M.); (M.C.C.)
| | - Erika Cortés-Macías
- Instituto de Agroquímica y Tecnología de los Alimentos (IATA-CSIC), 46980 Paterna, Spain; (C.B.); (E.C.-M.); (M.C.C.)
| | - Silvia Sánchez-González
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.M.); (A.O.-H.); (S.S.-G.)
| | | | - Julián Pérez-Villacastín
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Servicio de Cardiología, Hospital Clínico de San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.G.); (M.A.R.-C.)
- Departamento de Medicina, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
- Fundación para la Investigación Interhospitalaria Cardiovascular, 28008 Madrid, Spain
| | - María Carmen Collado
- Instituto de Agroquímica y Tecnología de los Alimentos (IATA-CSIC), 46980 Paterna, Spain; (C.B.); (E.C.-M.); (M.C.C.)
| | - Dulcenombre Gómez-Garre
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain; (J.M.); (A.O.-H.); (S.S.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain
| |
Collapse
|
14
|
Xiong HH, Lin SY, Chen LL, Ouyang KH, Wang WJ. The Interaction between Flavonoids and Intestinal Microbes: A Review. Foods 2023; 12:foods12020320. [PMID: 36673411 PMCID: PMC9857828 DOI: 10.3390/foods12020320] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/27/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
In recent years, research on the interaction between flavonoids and intestinal microbes have prompted a rash of food science, nutriology and biomedicine, complying with future research trends. The gut microbiota plays an essential role in the maintenance of intestinal homeostasis and human health, but once the intestinal flora dysregulation occurs, it may contribute to various diseases. Flavonoids have shown a variety of physiological activities, and are metabolized or biotransformed by gut microbiota, thereby producing new metabolites that promote human health by modulating the composition and structure of intestinal flora. Herein, this review demonstrates the key notion of flavonoids as well as intestinal microbiota and dysbiosis, aiming to provide a comprehensive understanding about how flavonoids regulate the diseases by gut microbiota. Emphasis is placed on the microbiota-flavonoid bidirectional interaction that affects the metabolic fate of flavonoids and their metabolites, thereby influencing their metabolic mechanism, biotransformation, bioavailability and bioactivity. Potentially by focusing on the abundance and diversity of gut microbiota as well as their metabolites such as bile acids, we discuss the influence mechanism of flavonoids on intestinal microbiota by protecting the intestinal barrier function and immune system. Additionally, the microbiota-flavonoid bidirectional interaction plays a crucial role in regulating various diseases. We explain the underlying regulation mechanism of several typical diseases including gastrointestinal diseases, obesity, diabetes and cancer, aiming to provide a theoretical basis and guideline for the promotion of gastrointestinal health as well as the treatment of diseases.
Collapse
Affiliation(s)
- Hui-Hui Xiong
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Su-Yun Lin
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Ling-Li Chen
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Ke-Hui Ouyang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Wen-Jun Wang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
- Correspondence: ; Tel.: +86-791-83813655
| |
Collapse
|
15
|
Zhang X, Zuo L, Geng Z, Song X, Li J, Ge S, Jiang Y, Yang Z, Liu G, Zhao Y, Zhao H, Yu L, Hu J. Vindoline ameliorates intestinal barrier damage in Crohn's disease mice through MAPK signaling pathway. FASEB J 2022; 36:e22589. [PMID: 36197455 DOI: 10.1096/fj.202200234rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 08/09/2022] [Accepted: 09/23/2022] [Indexed: 11/11/2022]
Abstract
Intestinal inflammation and intestinal barrier damage are important pathological changes in Crohn's disease (CD). Vindoline is a natural monomer with anti-inflammatory effects. We employed CD model mice to explore the effect of Vindoline on CD-like colitis and the possible mechanism. Il-10-deficient (Il-10-/- ) mice and wild-type (WT) mice (both aged 15 weeks, male) were used to explore the effect of Vindoline on colitis and intestinal barrier damage, as well as macrophage-mediated inflammation. Bone-marrow-derived macrophages (BMDMs) and colonic organoids from mice were used to explore the inhibitory effect of Vindoline on macrophage-mediated inflammation and the protective effect on inflammation-induced intestinal barrier damage as well as the possible mechanism. We found that Vindoline significantly ameliorated colitis in CD mice, as evidenced by increased weight change and colon length and decreased the colon macroscopic injury score, histological inflammatory score, and the expression of pro-inflammatory mediators. Vindoline also protected against intestinal barrier damage in CD mice. Furthermore, Vindoline inhibited macrophage-mediated inflammation and protected against inflammation-induced intestinal barrier damage in the coculture system. In addition, Vindoline ameliorated colitis in CD mice by protecting against inflammation-induced intestinal barrier damage, which may be caused by inhibition of MAPK signaling pathway. This protective effect suggests that Vindoline has potential value for clinical application in the treatment of CD.
Collapse
Affiliation(s)
- Xiaofeng Zhang
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Lugen Zuo
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhijun Geng
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Xue Song
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Jing Li
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Sitang Ge
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yifan Jiang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Zi Yang
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Guangyong Liu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Yajing Zhao
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Hao Zhao
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Liang Yu
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jianguo Hu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
16
|
Zhao J, Lin Z, Ying P, Zhao Z, Yang H, Qian J, Gong Y, Zhou Y, Dai Y, Jiao Y, Zhu W, Wang H, Tang L. circSMAD4 promotes experimental colitis and impairs intestinal barrier functions by targeting JAK2 through sponging miR-135a-5p. J Crohns Colitis 2022; 17:593-613. [PMID: 36239525 DOI: 10.1093/ecco-jcc/jjac154] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Numerous studies have explored the association between circular RNAs (circRNAs) and Crohn's disease (CD). However, the pathological role, biological functions, and molecular mechanisms of circRNAs in CD have not been fully elucidated. METHODS The circRNA microarray analysis was performed to identify deregulated circRNAs in colon tissues. The identified circRNA were verified through quantitative real time-polymerase chain reaction (qRT-PCR). In vivo and in vitro functional studies were performed to verify the role of circSMAD4 in CD and investigate the mechanisms involved. RESULTS We found that circSMAD4 was the most significantly upregulated circRNA. The expression level of circSMAD4 was positively correlated with levels of inflammatory factors. Overexpression of circSMAD4 impaired tight junction (TJ) proteins and enhanced apoptosis of epithelial cells. These effects were reversed by treatment with miR-135a-5p mimic. Mechanistic studies showed that circSMAD4 exerts its effects on CD by "sponging" miR-135a-5p to regulate Janus kinase 2 (JAK2). Si-circSMAD4 delivery through microspheres ameliorated experimental colitis and protected the intestinal barrier function in IL-10 knock-out mice. CONCLUSION This study shows that circSMAD4 regulates the progression of experimental colitis via the miR-135a-5p/JAK2 signaling axis and it may be a potential therapeutic target.
Collapse
Affiliation(s)
- Jie Zhao
- Department of Gastrointestinal Surgery and and Central Laboratory, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University
| | - Zhiliang Lin
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University
| | - Pu Ying
- Department of Orthopedics, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine
| | - Zhibin Zhao
- Department of Gastroenterology, Taizhou People's Hospital Affiliated to Nanjing Medical University
| | - Haojun Yang
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University
| | - Jun Qian
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University
| | - Yu Gong
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University
| | - Yan Zhou
- Department of Gastrointestinal Surgery and and Central Laboratory, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University
| | - Yi Dai
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University
| | - Yuwen Jiao
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University
| | - Weiming Zhu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University
| | - Honggang Wang
- Department of General Surgery, Taizhou People's Hospital Affiliated to Nanjing Medical University
| | - Liming Tang
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University
| |
Collapse
|
17
|
Bouabsa F, Tir Touil A, Al Zoubi MS, Chelli N, Leke A, Meddah B. Caffeine citrate effects on gastrointestinal permeability, bacterial translocation and biochemical parameters in newborn rats after long-term oral administration. MEDITERRANEAN JOURNAL OF NUTRITION AND METABOLISM 2022; 15:307-321. [DOI: 10.3233/mnm-211544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Caffeine is a potent central and respiratory acting agent used in neonatology to treat apnea in premature newborns. OBJECTIVE: This study investigates the effects of caffeine orally administered to newborn rats on gastrointestinal permeability, bacterial translocation and different biochemical parameters. METHODS: Newborn rats were divided into different groups ( N = 06). The treated newborn rats were orally administered with standard caffeine doses (12 mg/kg per day), and the control groups received a placebo. The animals were weighed daily until sacrifice. Blood samples, mesenteric lymph nodes (MLN) and organs were aseptically collected. Furthermore, different biochemical (D-Lactate) and oxidative stress biomarkers (MDA, CAT, SOD and GSH) were examined. Microbiological analyses were performed to assess microbiota alterations and bacterial translocation. RESULTS: Preliminary results showed that caffeine administration decreased the level of bacterial translocation over time. The treatment reduced plasma D-lactate levels ( p < 0.05). Additionally, caffeine induced a disturbance in the concentrations of biochemical parameters and oxidative stress biomarkers. Indeed, liver enzymes (AST and ALT) were significantly ( p < 0.05) risen after caffeine treatment. Glutathione (GSH) levels were significantly higher in caffeine treated groups (75.12±0.32; 51.98±1.12 U/mg; p < 0.05) comparing to control ones (40.82±0.25; 42.91±0.27 U/mg; p < 0.05) in the ileum and the colon, respectively. CONCLUSIONS: Thus, besides improving gastrointestinal permeability, our data show that caffeine has beneficial effects on the intestinal antioxidant system.
Collapse
Affiliation(s)
- Foufa Bouabsa
- Bioconversion, Microbiology Engineering and Health Safety Laboratory (LBGMSS), Nature and Life Sciences Faculty, Mustapha Stambouli University, Mascara, Algeria
| | - Aicha Tir Touil
- Bioconversion, Microbiology Engineering and Health Safety Laboratory (LBGMSS), Nature and Life Sciences Faculty, Mustapha Stambouli University, Mascara, Algeria
| | - Mazhar Salim Al Zoubi
- Department of Basic Medical Sciences, Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Nadia Chelli
- Bioconversion, Microbiology Engineering and Health Safety Laboratory (LBGMSS), Nature and Life Sciences Faculty, Mustapha Stambouli University, Mascara, Algeria
| | - André Leke
- Pediatric Neonatal Department, CHU Nord-Amiens, France
| | - Boumediene Meddah
- Bioconversion, Microbiology Engineering and Health Safety Laboratory (LBGMSS), Nature and Life Sciences Faculty, Mustapha Stambouli University, Mascara, Algeria
| |
Collapse
|
18
|
Chen J, Wang Y, Shi Y, Liu Y, Wu C, Luo Y. Association of Gut Microbiota With Intestinal Ischemia/Reperfusion Injury. Front Cell Infect Microbiol 2022; 12:962782. [PMID: 35903197 PMCID: PMC9314564 DOI: 10.3389/fcimb.2022.962782] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Intestinal ischemia/reperfusion (II/R) is a common acute and critical condition in clinical practice with a high mortality rate. However, there is still a lack of effective prevention and treatment measures for II/R injury. The role of the gut microbiota in II/R has attracted widespread attention. Recent evidence has demonstrated that the gut microbiota plays a pivotal role in the occurrence, development, and prognosis of II/R. Therefore, maintaining the homeostasis of gut microbiota and its metabolites may be a potential strategy for the treatment of II/R. This review focuses on the importance of crosstalk between the gastrointestinal ecosystem and II/R to highlight II/R-induced gut microbiota signatures and potential applications of microbial-based therapies in II/R. This will also provide potentially effective biomarkers for the prediction, diagnosis and treatment of II/R.
Collapse
Affiliation(s)
- Jingyi Chen
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- *Correspondence: Jingyi Chen, ; Yanrong Luo, ; Chengyi Wu,
| | - Yu Wang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yongxia Shi
- Department of Surgical Nursing, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yongpan Liu
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Chengyi Wu
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- *Correspondence: Jingyi Chen, ; Yanrong Luo, ; Chengyi Wu,
| | - Yanrong Luo
- Physical Examination Center, Shiyan Hospital of Integrated Traditional and Western Medicine, Shiyan, China
- *Correspondence: Jingyi Chen, ; Yanrong Luo, ; Chengyi Wu,
| |
Collapse
|
19
|
Lei J, Xie Y, Sheng J, Song J. Intestinal microbiota dysbiosis in acute kidney injury: novel insights into mechanisms and promising therapeutic strategies. Ren Fail 2022; 44:571-580. [PMID: 35350960 PMCID: PMC8967199 DOI: 10.1080/0886022x.2022.2056054] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In recent years, the clinical impact of intestinal microbiota–kidney interaction has been emerging. Experimental evidence highlighted a bidirectional evolutionary correlation between intestinal microbiota and kidney diseases. Nonetheless, acute kidney injury (AKI) is still a global public health concern associated with high morbidity, mortality, healthcare costs, and limited efficient therapy. Several studies on the intestinal microbiome have improved the knowledge and treatment of AKI. Therefore, the present review outlines the concept of the gut–kidney axis and data about intestinal microbiota dysbiosis in AKI to improve the understanding of the mechanisms of the intestinal microbiome on the modification of kidney function and response to kidney injury. We also introduced the future directions and research areas, emphasizing the intervention approaches and recent research advances of intestinal microbiota dysbiosis during AKI, thereby providing a new perspective for future clinical trials.
Collapse
Affiliation(s)
- Juan Lei
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yifan Xie
- Department of Rheumatism and Immunology, Children's Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Jingyi Sheng
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Jiayu Song
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
20
|
Suslov AV, Chairkina E, Shepetovskaya MD, Suslova IS, Khotina VA, Kirichenko TV, Postnov AY. The Neuroimmune Role of Intestinal Microbiota in the Pathogenesis of Cardiovascular Disease. J Clin Med 2021; 10:1995. [PMID: 34066528 PMCID: PMC8124579 DOI: 10.3390/jcm10091995] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/19/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
Currently, a bidirectional relationship between the gut microbiota and the nervous system, which is considered as microbiota-gut-brain axis, is being actively studied. This axis is believed to be a key mechanism in the formation of somatovisceral functions in the human body. The gut microbiota determines the level of activation of the hypothalamic-pituitary system. In particular, the intestinal microbiota is an important source of neuroimmune mediators in the pathogenesis of cardiovascular disease. This review reflects the current state of publications in PubMed and Scopus databases until December 2020 on the mechanisms of formation and participation of neuroimmune mediators associated with gut microbiota in the development of cardiovascular disease.
Collapse
Affiliation(s)
- Andrey V. Suslov
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, 8-2 Trubetskaya Str., 119992 Moscow, Russia; (A.V.S.); (E.C.); (M.D.S.)
| | - Elizaveta Chairkina
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, 8-2 Trubetskaya Str., 119992 Moscow, Russia; (A.V.S.); (E.C.); (M.D.S.)
| | - Maria D. Shepetovskaya
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, 8-2 Trubetskaya Str., 119992 Moscow, Russia; (A.V.S.); (E.C.); (M.D.S.)
| | - Irina S. Suslova
- Central State Medical Academy of the Administrative Department of the President of the Russian Federation, 19-1A Marshal Timoshenko Str., 121359 Moscow, Russia;
| | - Victoria A. Khotina
- Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (A.Y.P.)
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Str., 125315 Moscow, Russia
| | - Tatiana V. Kirichenko
- Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (A.Y.P.)
- National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Str., 121552 Moscow, Russia
| | - Anton Y. Postnov
- Research Institute of Human Morphology, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (A.Y.P.)
- National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Str., 121552 Moscow, Russia
| |
Collapse
|
21
|
Shin SY, Hussain Z, Lee YJ, Park H. An altered composition of fecal microbiota, organic acids, and the effect of probiotics in the guinea pig model of postoperative ileus. Neurogastroenterol Motil 2021; 33:e13966. [PMID: 32815235 DOI: 10.1111/nmo.13966] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/15/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND The aim of this study is to investigate the altered composition of fecal microbiota, organic acids, and the effect of probiotics in the guinea pig model of the postoperative ileus (POI). METHODS A laparotomy with cecal manipulation was performed to induce POI in guinea pigs. Fecal pellets were collected before the operation (the baseline) and 1, 3, and 5 days after the operation. The extracted fecal DNA was amplified and sequenced using the Illumina MiSeq sequencing system. The same POI procedures were performed after oral pretreatment of the probiotics for 7 days before operation. The effect of the probiotics on the selected taxa and fecal acetate were evaluated, as were the butyrate levels. The colonic transit was assessed by measurement of the fecal pellet output. KEY RESULTS The communities of the baseline and POI groups indicated significantly distinct composition. The genera Bifidobacterium and Lactobacillus were more abundant in the baseline group compared with the POI groups, and Bacteroides and Blautia were more abundant in the POI groups. Decreased abundances of the species Bifidobacterium bifidum and Bifidobacterium longum after the POI procedure were significantly increased in the probiotics group. The decreased fecal butyrate level after the POI procedure was significantly increased, and colonic transit was significantly improved in the probiotics group. CONCLUSIONS AND INFERENCES POI induces gut bacterial dysbiosis. Moreover, pretreatment of probiotics before operation restores the beneficial bacterial species, butyrate production, and bowel movement. The modulation of gut microbiota may help the treatment and prevention of POI.
Collapse
Affiliation(s)
- Seung Yong Shin
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Zahid Hussain
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young Ju Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyojin Park
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
22
|
Toxicity, gut microbiota and metabolome effects after copper exposure during early life in SD rats. Toxicology 2020; 433-434:152395. [PMID: 32027963 DOI: 10.1016/j.tox.2020.152395] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/20/2020] [Accepted: 02/02/2020] [Indexed: 12/27/2022]
Abstract
Copper, an essential microelement, can still be harmful to health and has a significant impact on the gut microbiota, which is closely related to health when copper is ingested excessively. However, the effects of low dose exposure to copper early in life on health and the gut microbiota are not well understood. Here, the effects of early-life exposure of copper on the toxicity, gut microbiota and the metabolome were investigated in Sprague-Dawley (SD) rats. The results showed that 0.20 and 1.00 mg/kg BW copper early-life exposure in SD rats significantly increased ALT, AST, and ALP levels in the blood and caused liver damage. Copper exposure had a dose-dependent effect on the alpha and beta diversity and reduced the abundance of probiotics, the ratio of Firmicutes to Bacteroidetes (F/B), and changed the abundance of fat metabolism and intestinal inflammation-related bacteria. The results of the fecal metabolome also demonstrated the effects of early-life copper exposure on liver damage and intestinal inflammation-related metabolic pathways. Together, our findings demonstrated that copper exposure during early life induced liver damage and gut microbiota dysbiosis and affected the relevant metabolic pathways.
Collapse
|
23
|
Abstract
OBJECTIVE In kidney diseases, uncontrolled blood pressure, inflammation, oxidative stress, imbalanced immunity response, and metabolic dysfunction were associated with the progressive deterioration of renal function. Short-chain fatty acids (SCFAs), as a group of metabolites fermented by gut microbiota exerted regulatory effects on kidney diseases through their activation of trans-membrane G protein-coupled receptors and their inhibition of histone acetylation. In this review article, we updated recent research advances that provided an opportunity to explore our understanding in physiology and function of SCFAs in kidney disease. DATA SOURCES We performed a comprehensive search in both PubMed and Embase using "short-chain fatty acids" and "kidney" with no restrictions on publication date. STUDY SELECTION After reading through the title and abstract for early screening, the full text of relevant studies was identified and reviewed to summarize the roles of SCFAs in kidney diseases. RESULTS Though controversial, growing evidence suggested SCFAs appeared to have a complex but yet poorly understood communications with cellular and molecular processes that affected kidney function and responses to injury. From recent studies, SCFAs influenced multiple aspects of renal physiology including inflammation and immunity, fibrosis, blood pressure, and energy metabolism. CONCLUSIONS The roles of intestinal SCFAs in kidney diseases were exciting regions in recent years; however, clinical trials and animal experiments in kidney diseases were still lacked. Thus, more research would be needed to obtain better understanding of SCFAs' potential effects in kidney diseases.
Collapse
|
24
|
Qin SM, Zhang KY, Ding XM, Bai SP, Wang JP, Zeng QF. Effect of dietary graded resistant potato starch levels on growth performance, plasma cytokines concentration, and intestinal health in meat ducks. Poult Sci 2019; 98:3523-3532. [PMID: 31329991 DOI: 10.3382/ps/pez186] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/18/2019] [Indexed: 12/15/2022] Open
Abstract
The objective of the present study was to investigate the effect of dietary graded raw potato starch (RPS) levels on growth performance, plasma cytokines concentration, ileal barrier function, and cecal short-chain fatty acids (SCFA) concentration in meat ducks from 1 to 35 D of age. This study included 2 experiments. In experiment (Exp.) 1, sixteen 35-day-old meat ducks were used to evaluate the AME of RPS by orogastric administration. Results showed the AME value of RPS on ducks is 2.76 kcal/g. In Exp. 2, a total of 600 one-day-old ducklings were randomly assigned to 5 isonitrogenous and isoenergetic dietary treatments that included 0 (control), 6, 12, 18, and 24% RPS, respectively. Samples were collected at both of 14 and 35 D. Neither growth performance nor ileal parameters (length, weight, and pH) at both of 14 and 35 D was affected by dietary RPS. However, the mucosal thickness (14 D), villus height (except for 18% RPS at 14 D), and the villus height: crypt depth ratio (14 and 35 D) of the ileum were increased in the 12 and 18% RPS diets when compared to 0% RPS diet. Meanwhile, proinflammatory factors such as plasma interleukin (IL)-1β and IL-6 (14 D) reduced in 12% RPS diet and tumor necrosis factor α decreased in 12% (except for 14 D) and 18% RPS groups. When compared with the control group, diets with 18% RPS significantly increased mucin 2 gene expression at 14 D, and 12% RPS elevated the mRNA expression of tight junction proteins including Zonula occludens-1 and Claudin 1 (except for 14 D) in the ileal mucosa of birds. Furthermore, ducks fed 12% RPS diet had higher concentrations of acetate, propionate, and butyrate in cecal digesta than other groups. These findings indicated that diets with 12 and/or 18% RPS increased the cecal SCFA concentration, which subsequently enhanced the barrier function and improved intestinal health in the ileum for 14 and 35-day-old meat ducks.
Collapse
Affiliation(s)
- S M Qin
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - K Y Zhang
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - X M Ding
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - S P Bai
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - J P Wang
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Q F Zeng
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| |
Collapse
|
25
|
Song X, Li J, Wang Y, Zhou C, Zhang Z, Shen M, Xiang P, Zhang X, Zhao H, Yu L, Zuo L, Hu J. Clematichinenoside AR ameliorated spontaneous colitis in Il-10 -/- mice associated with improving the intestinal barrier function and abnormal immune responses. Life Sci 2019; 239:117021. [PMID: 31678552 DOI: 10.1016/j.lfs.2019.117021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/21/2019] [Accepted: 10/26/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Clematichinenoside AR (AR) is a saponin extracted for traditional Chinese medicine with the effects of improving the expression of tight junction (TJ) proteins and mediating anti-inflammatory activities. However, its effect on Crohn's disease (CD) is still unknown. We aimed to investigate the impact of AR on CD-like colitis and determine the mechanism underlying its effects. METHODS Interleukin-10 gene knockout (Il-10-/-) mice (male, fifteen weeks old) with spontaneous colitis were allocated to the positive control and AR-treated (32 mg/kg AR administered every other day by gavage for 4 weeks) groups. Wild-type (WT) mice (male, fifteen weeks old) composed the negative control group. The effects of AR on intestinal barrier function and structure and T cell responses as well as the potential mechanisms underlying these effects were investigated. RESULTS AR treatment significantly improved spontaneous colitis in Il-10-/- mice as demonstrated by reductions in the inflammatory score, disease activity index (DAI) and levels of inflammatory factors. The effects of AR on colitis in Il-10-/- mice were related to protecting intestinal barrier function and maintaining immune system homeostasis (regulatory T cell (Treg)/T helper 17 (Th17) cell balance). The anticolitis effect of AR may partly act by downregulating PI3K/Akt signaling. CONCLUSIONS AR may have therapeutic potential for treating CD in humans.
Collapse
Affiliation(s)
- Xue Song
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Jing Li
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yan Wang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Clinical Medicine, Bengbu Medical College, Bengbu, China
| | - Changmin Zhou
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Clinical Medicine, Bengbu Medical College, Bengbu, China
| | - Zhichao Zhang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Clinical Medicine, Bengbu Medical College, Bengbu, China
| | - Mengdi Shen
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Clinical Medicine, Bengbu Medical College, Bengbu, China
| | - Ping Xiang
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Xiaofeng Zhang
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Hao Zhao
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Liang Yu
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Lugen Zuo
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Jianguo Hu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China; Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.
| |
Collapse
|
26
|
Regulation of Gut Microbiota and Metabolic Endotoxemia with Dietary Factors. Nutrients 2019; 11:nu11102277. [PMID: 31547555 PMCID: PMC6835897 DOI: 10.3390/nu11102277] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/13/2019] [Accepted: 09/18/2019] [Indexed: 02/08/2023] Open
Abstract
Metabolic endotoxemia is a condition in which blood lipopolysaccharide (LPS) levels are elevated, regardless of the presence of obvious infection. It has been suggested to lead to chronic inflammation-related diseases such as obesity, type 2 diabetes mellitus, non-alcoholic fatty liver disease (NAFLD), pancreatitis, amyotrophic lateral sclerosis, and Alzheimer’s disease. In addition, it has attracted attention as a target for the prevention and treatment of these chronic diseases. As metabolic endotoxemia was first reported in mice that were fed a high-fat diet, research regarding its relationship with diets has been actively conducted in humans and animals. In this review, we summarize the relationship between fat intake and induction of metabolic endotoxemia, focusing on gut dysbiosis and the influx, kinetics, and metabolism of LPS. We also summarize the recent findings about dietary factors that attenuate metabolic endotoxemia, focusing on the regulation of gut microbiota. We hope that in the future, control of metabolic endotoxemia using dietary factors will help maintain human health.
Collapse
|
27
|
Ahmad AF, Dwivedi G, O'Gara F, Caparros-Martin J, Ward NC. The gut microbiome and cardiovascular disease: current knowledge and clinical potential. Am J Physiol Heart Circ Physiol 2019; 317:H923-H938. [PMID: 31469291 DOI: 10.1152/ajpheart.00376.2019] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. The human body is populated by a diverse community of microbes, dominated by bacteria, but also including viruses and fungi. The largest and most complex of these communities is located in the gastrointestinal system and, with its associated genome, is known as the gut microbiome. Gut microbiome perturbations and related dysbiosis have been implicated in the progression and pathogenesis of CVD, including atherosclerosis, hypertension, and heart failure. Although there have been advances in the characterization and analysis of the gut microbiota and associated bacterial metabolites, the exact mechanisms through which they exert their action are not well understood. This review will focus on the role of the gut microbiome and associated functional components in the development and progression of atherosclerosis. Potential treatments to alter the gut microbiome to prevent or treat atherosclerosis and CVD are also discussed.
Collapse
Affiliation(s)
- Adilah F Ahmad
- Medical School, University of Western Australia, Perth, Western Australia, Australia.,Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
| | - Girish Dwivedi
- Medical School, University of Western Australia, Perth, Western Australia, Australia.,Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia.,Department of Cardiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Fergal O'Gara
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia, Australia.,Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,BIOMERIT Research Centre, School of Microbiology, University College Cork, National University of Ireland, Cork, Ireland.,Telethon Kids Institute, Children's Hospital, Perth, Western Australia, Australia
| | - Jose Caparros-Martin
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth, Western Australia, Australia.,Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Natalie C Ward
- Medical School, University of Western Australia, Perth, Western Australia, Australia.,School of Public Health, Curtin University, Perth Western Australia, Australia
| |
Collapse
|
28
|
姜 从, 朱 平, 时 依, 项 武, 葛 思, 张 宗, 左 芦. [Protective effect of procyanidin B2 on intestinal barrier and against enteritis in a mouse model of trinitrobenzene sulphonic acid-induced colitis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:778-783. [PMID: 31340909 PMCID: PMC6765556 DOI: 10.12122/j.issn.1673-4254.2019.07.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the protective effect of procyanidin B2 (PCB2) on the intestinal barrier and against enteritis in mice with trinitrobenzene sulphonic acid (TNBS)-induced colitis and explore the possible mechanism. METHODS A mouse model of TNBS-induced colitis was established in male Balb/c mice aged 6-8 weeks. The successfully established mouse models were randomly divided into PCB2 treatment group (n=10) and model group (n=10) and were treated with daily intragastric administration of PCB2 (100 mg/kg, 0.2 mL) and 0.2 mL normal saline, respectively. After 4 weeks, the disease symptoms, intestinal inflammation, intestinal mucosal cell barrier function and the changes in PI3K/AKT signaling were evaluated using HE staining, immunofluorescence assay and Western blotting. RESULTS The disease activity index of the mice was significantly lower and the mean body weight was significantly greater in PCB2 group than in the model group in the 3rd and 4th weeks of intervention (P < 0.05). The levels of colonic inflammation and intestinal mucosal inflammatory mediators IL-1β and TNF-α were significantly lower while IL-10 was significantly higher in PCB2 group than in the model group (P < 0.05). Compared with those in the model group, the mice in PCB2 treatment group showed a significantly lower positive rate of bacterial translocation in the mesenteric lymph nodes and a lower thiocyanate-dextran permeability of the intestinal mucosa (P < 0.05). Western blotting showed that PCB2 treatment significantly increased the expressions of claudin-1 and ZO-1 (P < 0.05) and significantly lowered the expression levels of p-PI3K and p-AKT in the intestinal mucosa as compared with those in the model group (P < 0.05). CONCLUSIONS PCB2 suppresses intestinal inflammation and protects intestinal mucosal functions and structural integrity by inhibiting intestinal PI3K/AKT signaling pathway, suggesting the potential of PCB2 as a new drug for Crohn's disease.
Collapse
Affiliation(s)
- 从桥 姜
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 平胜 朱
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 依 时
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 武军 项
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 思堂 葛
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 宗兵 张
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - 芦根 左
- />蚌埠医学院第一附属医院胃肠外科,安徽 蚌埠 233004Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
29
|
Zuo L, Li J, Ge S, Ge Y, Shen M, Wang Y, Zhou C, Wu R, Hu J. Bryostatin-1 ameliorated experimental colitis in Il-10 -/- Mice by protecting the intestinal barrier and limiting immune dysfunction. J Cell Mol Med 2019; 23:5588-5599. [PMID: 31251471 PMCID: PMC6652299 DOI: 10.1111/jcmm.14457] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/13/2022] Open
Abstract
Bryostatin‐1 (Bry‐1) has been proven to be effective and safe in clinical trials of a variety of immune‐related diseases. However, little is known about its effect on Crohn's disease (CD). We aimed to investigate the impact of Bry‐1 on CD‐like colitis and determine the mechanism underlying this effect. In the present study, 15‐week‐old male Il‐10−/− mice with spontaneous colitis were divided into positive control and Bry‐1‐treated (Bry‐1, 30 μg/kg every other day, injected intraperitoneally for 4 weeks) groups. Age‐matched, male wild‐type (WT) mice were used as a negative control. The effects of Bry‐1 on colitis, intestinal barrier function and T cell responses as well as the potential regulatory mechanisms were evaluated. We found that the systemic delivery of Bry‐1 significantly ameliorated colitis in Il‐10−/− mice, as demonstrated by decreases in the disease activity index (DAI), inflammatory score and proinflammatory mediator levels. The protective effects of Bry‐1 on CD‐like colitis included the maintenance of intestinal barrier integrity and the helper T cell (Th)/regulatory T cell (Treg) balance. These effects of Bry‐1 may act in part through nuclear factor erythroid 2‐related factor 2 (Nrf2) signalling activation and STAT3/4 signalling inhibition. The protective effect of Bry‐1 on CD‐like colitis suggests Bry‐1 has therapeutic potential in human CD, particularly given the established clinical safety of Bry‐1.
Collapse
Affiliation(s)
- Lugen Zuo
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Jing Li
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Sitang Ge
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Yuanyuan Ge
- Department of Colorectal Surgery, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Mengdi Shen
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Clinical medicine, Bengbu medical college, Bengbu, China
| | - Yan Wang
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Clinical medicine, Bengbu medical college, Bengbu, China
| | - Changmin Zhou
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Clinical medicine, Bengbu medical college, Bengbu, China
| | - Rong Wu
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jianguo Hu
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
30
|
Li J, Zuo L, Tian Y, He Y, Zhang Z, Guo P, Ge Y, Hu J. Spontaneous colitis in IL-10-deficient mice was ameliorated via inhibiting glutaminase1. J Cell Mol Med 2019; 23:5632-5641. [PMID: 31211512 PMCID: PMC6653008 DOI: 10.1111/jcmm.14471] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/09/2019] [Accepted: 04/26/2019] [Indexed: 12/30/2022] Open
Abstract
Immunity imbalance and barrier damage in the intestinal mucosa are the main pathogenic factors of Crohn's disease (CD). Bis‐2‐(5‐phenylacetamido‐1,2,4‐thiadiazol‐2‐yl) ethyl sulfide (BPTES) is a glutaminase 1 (Gls1) inhibitor with the dual functions of increasing glutamine levels and immune regulation. In this study, we focused on the role of BPTES in CD‐like enteritis and the possible mechanisms. We found that Gls1 expression was significantly increased in CD intestinal tissue compared with control tissue. Bis‐2‐(5‐phenylacetamido‐1,2,4‐thiadiazol‐2‐yl) ethyl sulfide treatment significantly ameliorated chronic colitis in the IL‐10−/−, as manifested by decreased disease activity index, body weight change, histological inflammatory degree and inflammatory cytokine expression. Bis‐2‐(5‐phenylacetamido‐1,2,4‐thiadiazol‐2‐yl) ethyl sulfide treatment exerted protective effects on CD that were associated with the maintenance of intestinal barrier integrity and the Th/Treg balance. Bis‐2‐(5‐phenylacetamido‐1,2,4‐thiadiazol‐2‐yl) ethyl sulfide treatment may act in part through TCR‐mediated mammalian target of rapamycin complex 1 (mTORC1) signalling activation. In conclusion, inhibition of Gls1 expression attenuated chronic colitis by maintaining intestinal barrier integrity and the Th/Treg balance, thereby ameliorating CD‐like colitis.
Collapse
Affiliation(s)
- Jing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Lugen Zuo
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China.,Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yun Tian
- Department of Oncology, Shanghai Dermatology Hospital, Tongji University, Shanghai, China.,Tongji University Cancer Center, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Yifan He
- Clinical Medicine of Bengbu Medical College, Bengbu, Anhui, China
| | - Zhichao Zhang
- Clinical Medicine of Bengbu Medical College, Bengbu, Anhui, China
| | - Pu Guo
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yuanyuan Ge
- Department of Colorectal Surgery, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianguo Hu
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.,Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
31
|
Cheng R, Guo J, Pu F, Wan C, Shi L, Li H, Yang Y, Huang C, Li M, He F. Loading ceftriaxone, vancomycin, and Bifidobacteria bifidum TMC3115 to neonatal mice could differently and consequently affect intestinal microbiota and immunity in adulthood. Sci Rep 2019; 9:3254. [PMID: 30824845 PMCID: PMC6397183 DOI: 10.1038/s41598-018-35737-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/18/2018] [Indexed: 02/05/2023] Open
Abstract
Recent studies have demonstrated that antibiotics/or probiotics administration in early life play key roles on modulating intestinal microbiota and the alterations might cause long-lasting consequences both physiologically and immunologically. We investigated the effects of early life ceftriaxone, vancomycin and Bifidobacterium bifidum TMC3115 (TMC3115) treatment on intestinal microbiota and immunity both in neonates and adults even after termination of antibiotics exposure. We found that ceftriaxone and vancomycin, but not TMC3115, significantly altered the intestinal microbiota, serum total IgE level, and the morphology and function of the intestinal epithelium in the neonatal mice. In the adult stages, the diversity and composition of the intestinal microbiota were significantly different in the antibiotic-treated mice, and ceftriaxone-treated mice exhibited significantly higher serum total IgE and OVA-specific IgE levels. TMC3115 significantly mitigated the alteration of intestinal microbiota caused by ceftriaxone not vancomycin. Antibiotics and TMC3115 can differently modulate intestinal microbiota and SCFAs metabolism, affecting the development and function of the immunity and intestinal epithelium to different degrees in neonatal mice. Neonatal ceftriaxone-induced abnormal intestinal microbiota, immunity and epithelium could last to adulthood partly, which might be associated with the enhancement of host susceptibility to IgE-mediated allergies and related immune responses, TMC3115 may protect against the side effects of antibiotic treatment, at least partly.
Collapse
Affiliation(s)
- RuYue Cheng
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, 610041, Chengdu, Sichuan, PR China
| | - JiaWen Guo
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, 610041, Chengdu, Sichuan, PR China
| | - FangFang Pu
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, 610041, Chengdu, Sichuan, PR China
| | - ChaoMin Wan
- Department of Pediatrics of Western China Second Hospital of Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, 610041, Chengdu, Sichuan, PR China
| | - Lei Shi
- Department of Clinical Nutrition, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, PR China
| | - HuaWen Li
- Hebei Inatural Biotech Co., Ltd, 050000, Shijiazhuang, Hebei, PR China
| | - YuHong Yang
- Hebei Inatural Biotech Co., Ltd, 050000, Shijiazhuang, Hebei, PR China
| | - ChengYu Huang
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, 610041, Chengdu, Sichuan, PR China
| | - Ming Li
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, 610041, Chengdu, Sichuan, PR China.
| | - Fang He
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, 610041, Chengdu, Sichuan, PR China.
| |
Collapse
|
32
|
Markey L, Shaban L, Green ER, Lemon KP, Mecsas J, Kumamoto CA. Pre-colonization with the commensal fungus Candida albicans reduces murine susceptibility to Clostridium difficile infection. Gut Microbes 2018; 9:497-509. [PMID: 29667487 PMCID: PMC6287688 DOI: 10.1080/19490976.2018.1465158] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/25/2018] [Accepted: 04/04/2018] [Indexed: 02/03/2023] Open
Abstract
Clostridium difficile is a major nosocomial pathogen responsible for close to half a million infections and 27,000 deaths annually in the U.S. Preceding antibiotic treatment is a major risk factor for C. difficile infection (CDI) leading to recognition that commensal microbes play a key role in resistance to CDI. Current antibiotic treatment of CDI is only partially successful due to a high rate of relapse. As a result, there is interest in understanding the effects of microbes on CDI susceptibility to support treatment of patients with probiotic microbes or entire microbial communities (e.g., fecal microbiota transplantation). The results reported here demonstrate that colonization with the human commensal fungus Candida albicans protects against lethal CDI in a murine model. Colonization with C. albicans did not increase the colonization resistance of the host. Rather, our findings showed that one effect of C. albicans colonization was to enhance a protective immune response. Mice pre-colonized with C. albicans expressed higher levels of IL-17A in infected tissue following C. difficile challenge compared to mice that were not colonized with C. albicans. Administration of cytokine IL-17A was demonstrated to be protective against lethal murine CDI in mice not colonized with C. albicans. C. albicans colonization was associated with changes in the abundance of some bacterial components of the gut microbiota. Therefore, C. albicans colonization altered the gut ecosystem, enhancing survival after C. difficile challenge. These findings demonstrate a new, beneficial role for C. albicans gut colonization.
Collapse
Affiliation(s)
- Laura Markey
- Graduate Program in Molecular Microbiology, Sackler School of Graduate Biomedical Sciences and Department of Molecular Biology and Microbiology, Tufts University, Boston, MA
| | - Lamyaa Shaban
- Graduate Program in Molecular Microbiology, Sackler School of Graduate Biomedical Sciences and Department of Molecular Biology and Microbiology, Tufts University, Boston, MA
| | - Erin R. Green
- Graduate Program in Molecular Microbiology, Sackler School of Graduate Biomedical Sciences and Department of Molecular Biology and Microbiology, Tufts University, Boston, MA
| | - Katherine P. Lemon
- The Forsyth Institute (Microbiology), Cambridge, MA and Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Joan Mecsas
- Graduate Program in Molecular Microbiology, Sackler School of Graduate Biomedical Sciences and Department of Molecular Biology and Microbiology, Tufts University, Boston, MA
| | - Carol A. Kumamoto
- Graduate Program in Molecular Microbiology, Sackler School of Graduate Biomedical Sciences and Department of Molecular Biology and Microbiology, Tufts University, Boston, MA
| |
Collapse
|
33
|
Duranti S, Vivo V, Zini I, Milani C, Mangifesta M, Anzalone R, Mancabelli L, Viappiani A, Cantoni AM, Barocelli E, van Sinderen D, Bertoni S, Turroni F. Bifidobacterium bifidum PRL2010 alleviates intestinal ischemia/reperfusion injury. PLoS One 2018; 13:e0202670. [PMID: 30161157 PMCID: PMC6116995 DOI: 10.1371/journal.pone.0202670] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 08/07/2018] [Indexed: 02/01/2023] Open
Abstract
Mesenteric ischemia/reperfusion is a clinical emergency with high morbidity and mortality due to the transient reduction of blood supply to the bowel. In recent years, the critical contribution of gut microbiome to human health and proper gastrointestinal functions has gradually emerged. In the current study, we investigated the protective effects of five days supplementation with Bifidobacterium bifidum PRL2010 in a murine model of gut ischemia/reperfusion. Our findings indicate that animals pretreated with B. bifidum PRL2010 showed lower neutrophil recruitment in the lungs, remarkably reduced bacterial translocation and decreased transcription levels of TNFalpha and IL-10 both in liver and kidneys, at the same time increasing those of IL-12 in kidneys. Inhibiting the adhesion of pathogenic bacteria and boosting host innate immunity responses are among the possible protective mechanisms enacted by the probiotic. These results demonstrate that short-period treatment with B. bifidum PRL2010 is a potential strategy to dampen remote organ injury due to mesenteric ischemia/reperfusion.
Collapse
Affiliation(s)
- Sabrina Duranti
- Laboratory of Probiogenomics, Department of Chemical Sciences, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Valentina Vivo
- Food and Drug Department, University of Parma, Parma, Italy
| | - Irene Zini
- Food and Drug Department, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemical Sciences, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | | | - Rosaria Anzalone
- Laboratory of Probiogenomics, Department of Chemical Sciences, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemical Sciences, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Alice Viappiani
- Laboratory of Probiogenomics, Department of Chemical Sciences, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | | | - Elisabetta Barocelli
- Laboratory of Probiogenomics, Department of Chemical Sciences, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, Cork, Ireland
| | - Simona Bertoni
- Food and Drug Department, University of Parma, Parma, Italy
- * E-mail:
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemical Sciences, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
34
|
Luo L, Zhou Z, Xue J, Wang Y, Zhang J, Cai X, Liu Y, Yang F. Bletilla striata polysaccharide has a protective effect on intestinal epithelial barrier disruption in TAA-induced cirrhotic rats. Exp Ther Med 2018; 16:1715-1722. [PMID: 30186392 PMCID: PMC6122099 DOI: 10.3892/etm.2018.6430] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/08/2018] [Indexed: 12/20/2022] Open
Abstract
It has been reported that intestinal epithelial barrier dysfunction serves an important role in the development of liver cirrhosis. However, at present there is no satisfactory treatment for intestinal mucosal lesions and ulcers associated with cirrhosis. The aim of the present study was to investigate the effect of Bletilla striata polysaccharide (BSP) on intestinal epithelial barrier disruption in rats with thioacetamide (TAA)-induced liver cirrhosis. Rats were randomly divided into 5 groups (n=10): BSP low dosage (15 mg/kg), BSP middle dosage (30 mg/kg), BSP high dosage (60 mg/kg), experiment and control groups. All groups except control group were administered with TAA (200 mg/kg/day) to induce liver cirrhosis. Following modeling, rats in the low, middle and high-dose BSP groups received BSP. ELISA kits were used to measure the endotoxin, alanine transaminase (ALT) and aspartate transaminase (AST) levels in the portal vein, while interleukin (IL)-6 and tumor necrosis factor (TNF)-α expression in the ileal tissue was measured. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting were used to detect the expression of zonula occludens (ZO)-1 and occludin mRNA and protein, respectively. Intestinal epithelial tissue pathology was detected using hematoxylin-eosin (HE) staining. Immunohistochemistry was performed to assess the expression of ZO-1 and occludin in intestinal epithelial tissues. Following treatment with BSP, ALT, AST and endotoxin levels in the portal vein, as well as IL-6 and TNF-α expression in ileal tissues, were significantly decreased compared with model group (P<0.05 or 0.01). Furthermore, BSP treatment upregulated the expression of ZO-1 and occludin mRNA and protein compared with the model group (P<0.05 or 0.01) and cytoplasmic staining for these proteins was increased. The degree of intestinal epithelial tissue pathological damage was significantly reduced in the BSP groups. In conclusion, BSP is able to reduce endotoxin levels, inhibit the inflammatory cytokines IL-6 and TNF-α and elevate expression of ZO-1 and occludin at tight junctions. Together, these results suggest a novel protective agent for the restoration of intestinal epithelial barrier disruption.
Collapse
Affiliation(s)
- Lei Luo
- Department of Gastroenterology, The Second People's Hospital of Yichang, Yichang, Hubei 443000, P.R. China
| | - Zhang Zhou
- Department of Anesthesia, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Juan Xue
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese and Western Medicine, Wuhan, Hubei 430015, P.R. China
| | - Yao Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Juan Zhang
- Department of Pulmonary Diseases, Jingmen City Hospital of Traditional Chinese Medicine, Jingmen, Hubei 448000, P.R. China
| | - Xin Cai
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan, Hubei 430060, P.R. China
| | - Yuqing Liu
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan, Hubei 430060, P.R. China
| | - Fan Yang
- Department of Hepatology, Hubei Provincial Hospital of Chinese Medicine, Wuhan, Hubei 430074, P.R. China
| |
Collapse
|
35
|
Bertoni S, Ballabeni V, Barocelli E, Tognolini M. Mesenteric ischemia-reperfusion: an overview of preclinical drug strategies. Drug Discov Today 2018; 23:1416-1425. [DOI: 10.1016/j.drudis.2018.05.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/27/2018] [Accepted: 05/24/2018] [Indexed: 02/06/2023]
|
36
|
Lei Q, Bi J, Chen H, Tian F, Gao X, Li N, Wang X. Glucagon-like peptide-2 improves intestinal immune function and diminishes bacterial translocation in a mouse model of parenteral nutrition. Nutr Res 2018; 49:56-66. [DOI: 10.1016/j.nutres.2017.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 09/29/2017] [Accepted: 10/05/2017] [Indexed: 02/07/2023]
|
37
|
Abstract
Gut microbiota and its metabolites play pivotal roles in host physiology and pathology. Short-chain fatty acids (SCFAs), as a group of metabolites, exert positive regulatory effects on energy metabolism, hormone secretion, immune inflammation, hypertension, and cancer. The functions of SCFAs are related to their activation of transmembrane G protein-coupled receptors and their inhibition of histone acetylation. Though controversial, growing evidence suggests that SCFAs, which regulate inflammation, oxidative stress, and fibrosis, have been involved in kidney disease through the activation of the gut–kidney axis; however, the molecular relationship among gut microbiota–derived metabolites, signaling pathways, and kidney disease remains to be elucidated. This review will provide an overview of the physiology and functions of SCFAs in kidney disease.
Collapse
Affiliation(s)
- Lingzhi Li
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Liang Ma
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ping Fu
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
38
|
Abstract
Surgery involving the gastrointestinal tract continues to prove challenging because of the persistence of unpredictable complications such as anastomotic leakage and life-threatening infections. Removal of diseased intestinal segments results in substantial catabolic stress and might require complex reconstructive surgery to maintain the functional continuity of the intestinal tract. As gastrointestinal surgery necessarily involves a breach of an epithelial barrier colonized by microorganisms, preoperative intestinal antisepsis is used to reduce infection-related complications. The current approach to intestinal antisepsis varies widely across institutions and countries with little understanding of its mechanism of action, effect on the gut microbiota and overall efficacy. Many of the current approaches to intestinal antisepsis before gastrointestinal surgery run counter to emerging concepts of intestinal microbiota contributing to immune function and recovery from injury. Here, we review evidence outlining the role of gut microbiota in recovery from gastrointestinal surgery, particularly in the development of infections and anastomotic leak. To make surgery safer and further reduce complications, a molecular, genetic and functional understanding of the response of the gastrointestinal tract to alterations in its microbiota is needed. Methods can then be developed to preserve the health-promoting functions of the microbiota while at the same time suppressing their harmful effects.
Collapse
Affiliation(s)
- Kristina Guyton
- MC-6040, Department of Surgery, University of Chicago Medicine, 5841 South Maryland Avenue, Chicago, Illinois 60637, USA
| | - John C Alverdy
- MC-6090, Department of Surgery, University of Chicago Medicine, 5841 South Maryland Avenue, Chicago, Illinois 60637, USA
| |
Collapse
|
39
|
Abstract
Ischemic disorders, such as myocardial infarction, stroke, and peripheral vascular disease, are the most common causes of debilitating disease and death in westernized cultures. The extent of tissue injury relates directly to the extent of blood flow reduction and to the length of the ischemic period, which influence the levels to which cellular ATP and intracellular pH are reduced. By impairing ATPase-dependent ion transport, ischemia causes intracellular and mitochondrial calcium levels to increase (calcium overload). Cell volume regulatory mechanisms are also disrupted by the lack of ATP, which can induce lysis of organelle and plasma membranes. Reperfusion, although required to salvage oxygen-starved tissues, produces paradoxical tissue responses that fuel the production of reactive oxygen species (oxygen paradox), sequestration of proinflammatory immunocytes in ischemic tissues, endoplasmic reticulum stress, and development of postischemic capillary no-reflow, which amplify tissue injury. These pathologic events culminate in opening of mitochondrial permeability transition pores as a common end-effector of ischemia/reperfusion (I/R)-induced cell lysis and death. Emerging concepts include the influence of the intestinal microbiome, fetal programming, epigenetic changes, and microparticles in the pathogenesis of I/R. The overall goal of this review is to describe these and other mechanisms that contribute to I/R injury. Because so many different deleterious events participate in I/R, it is clear that therapeutic approaches will be effective only when multiple pathologic processes are targeted. In addition, the translational significance of I/R research will be enhanced by much wider use of animal models that incorporate the complicating effects of risk factors for cardiovascular disease. © 2017 American Physiological Society. Compr Physiol 7:113-170, 2017.
Collapse
Affiliation(s)
- Theodore Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Christopher P. Baines
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Ronald J. Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
40
|
Wang H, Shi P, Zuo L, Dong J, Zhao J, Liu Q, Zhu W. Dietary Non-digestible Polysaccharides Ameliorate Intestinal Epithelial Barrier Dysfunction in IL-10 Knockout Mice. J Crohns Colitis 2016; 10:1076-86. [PMID: 26944415 DOI: 10.1093/ecco-jcc/jjw065] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Enteral nutrition [EN] was reported to be as effective as steroids in achieving short-term remission in patients with Crohn's disease [CD], and exclusive EN [EEN] is widely used as primary therapy in children with CD. The aim of this study was to investigate the effect of a specific multi-fibre mix [MF], designed to match the fibre content of a healthy diet, on intestinal epithelial barrier function in IL-10 knockout [IL-10(-/-)] mice with spontaneous chronic colitis. METHODS IL-10(-/-) mice aged 16 weeks, with established colitis, were used for the experiments with multi-fibre mix diet [MF] for 4 weeks. Severity of colitis, levels of short cahin fatty acids [SCFA] in caecum contents, expression of STAT 3 and STAT 4 proteins, CD4(+) CD45(+) lymphocytes, CD4(+)Foxp3(+) regulatory T cells [Tregs] and cytokines in the lamina propria [LP], epithelial expression of tight junction proteins, TNF-α/TNFR2 mRNA expression, and epithelial apoptosis in the proximal colon were measured at the end of the experiment. RESULTS MF feeding effectively attenuated disease activity index and colitis associated with decreased lamina propria CD4(+) CD45(+) lymphocytes, IFN-γ/IL-17A mRNA expression, and p-STAT 3 and p-STAT 4 expression in colonic mucosa of IL-10(-/-) mice [p < 0.05]. Furthermore, CD4(+)Foxp3(+) Tregs in the LP and concentrations of total SCFA, acetate, propionate, and butyrate in the caecum were markedly increased after MF feeding in IL-10(-/-) mice. After MF feeding, increased epithelial expression and correct localisation of tight junction proteins [occludin and zona occludens protein 1], as well as reduced TNF-α/TNFR2 mRNA expression and epithelial apoptosis, were also observed in IL-10(-/-) mice. CONCLUSIONS These results indicated that EEN supplemented with the tested fibre mix, known to modulate the intestinal microbiota composition and SCFA production, could possibly improve efficacy in inducing remission in patients with active CD.
Collapse
Affiliation(s)
- Honggang Wang
- Department of General Surgery, Taizhou People's Hospital, Medical School of Nantong University, Taizhou, China Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Peiliang Shi
- Model Animal Research Center of Nanjing University, Nanjing, China
| | - Lugen Zuo
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jianning Dong
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jie Zhao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qinghong Liu
- Department of General Surgery, Taizhou People's Hospital, Medical School of Nantong University, Taizhou, China
| | - Weiming Zhu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
41
|
Gut microbiota in renal physiology: focus on short-chain fatty acids and their receptors. Kidney Int 2016; 90:1191-1198. [PMID: 27575555 DOI: 10.1016/j.kint.2016.06.033] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 12/14/2022]
Abstract
A number of recent studies have begun to explore a new and exciting area: the interaction between the gut microbiome and renal physiology. In particular, multiple studies have focused on the role of microbially produced short chain fatty acids, which are generally thought to promote health. This review will focus on what is known to date regarding the influence of the microbiome on renal function, with emphasis on the cell biology, physiology, and clinical implications of short chain fatty acids and short chain fatty acid receptors. It is clear that microbe-host interactions are an exciting and ever-expanding field, which has implications for how we view diseases such as hypertension, acute kidney injury, and chronic kidney disease. However, it is important to recognize that although the potential promise of this area is extremely enticing, we are only the very edge of this new field.
Collapse
|
42
|
The microbiota and chronic kidney diseases: a double-edged sword. Clin Transl Immunology 2016; 5:e86. [PMID: 27757226 PMCID: PMC5067952 DOI: 10.1038/cti.2016.36] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/11/2016] [Accepted: 05/11/2016] [Indexed: 12/11/2022] Open
Abstract
Recent findings regarding the influence of the microbiota in many inflammatory processes have provided a new way to treat diseases. Now, one may hypothesize that the origin of a plethora of diseases is related to the health of the gut microbiota and its delicate, although complex, interface with the epithelial and immune systems. The ‘westernization' of diets, for example, is associated with alterations in the gut microbiota. Such alterations have been found to correlate directly with the increased incidence of diabetes and hypertension, the main causes of chronic kidney diseases (CKDs), which, in turn, have a high estimated prevalence. Indeed, data have arisen showing that the progression of kidney diseases is strictly related to the composition of the microbiota. Alterations in the gut microbiota diversity during CKDs do not only have the potential to exacerbate renal injury but may also contribute to the development of associated comorbidities, such as cardiovascular diseases and insulin resistance. In this review, we discuss how dysbiosis through alterations in the gut barrier and the consequent activation of immune system could intensify the progression of CKD and vice versa, how CKDs can modify the gut microbiota diversity and abundance.
Collapse
|
43
|
Effects ofl-carnitine and/or maize distillers dried grains with solubles in diets of gestating and lactating sows on the intestinal barrier functions of their offspring. Br J Nutr 2016; 116:459-69. [DOI: 10.1017/s0007114516001951] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AbstractThe objective of this study was to investigate the effects ofl-carnitine and/or maize distillers dried grains with solubles (DDGS) in diets of gestating and lactating sows on the intestinal barrier functions of their offspring. The experiment was designed as a 2×2 factorial with two dietary treatments (soyabean mealv.DDGS) and twol-carnitine levels (0v.100 mg/kg in gestating diets and 0v.200 mg/kg in lactating diets). Sows (Landrace×Large White) with an average parity of 4·2 with similar body weight were randomly assigned to four groups of thirty each. Dietary supplementation withl-carnitine increased the total superoxide dismutase activity but decreased the concentration of malondialdehyde of the jejunal mucosa in newborn piglets and weaning piglets on day 21. Dietary supplementation withl-carnitine decreased the concentrations of IL-1β, IL-12 and TNF-αin the jejunal mucosa of newborn piglets and decreased the concentrations of IL-6 and TNF-αin the jejunal mucosa of weaning piglets on day 21. There was an interaction between dietary treatment andl-carnitine on the bacterial numbers of total eubacteria in the digesta of caecum in weaning piglets on day 21. Bacterial numbers of total eubacteria in weaning piglets on day 21 were significantly increased byl-carnitine only in soyabean meal diet, but there was no significant effect ofl-carnitine in DDGS-based diet. Dietary supplementation withl-carnitine increased the bacterial numbers ofLactobacillusspp. and bifidobacteria spp. in the digesta of caecum in weaning piglets on day 21. Dietary supplementation withl-carnitine in sows affected the expression of tight junction proteins (claudin 1, zonula occludens-1 (ZO-1) and occludin) in the jejunal mucosa of their offspring by increasing the expression of ZO-1 mRNA in the jejunal mucosa of newborn piglets, and by increasing the expression of ZO-1 and occludin mRNA in the jejunal mucosa of weaning piglets on day 21. In conclusion, dietary supplementation withl-carnitine in gestating and lactating sows had positive effects on intestinal barrier functions of newborn piglets and weaning piglets on day 21, but it did not have effects on intestinal barrier functions of growing–finishing pigs in the filial generation. There were no effects of dietary treatment of sows on intestinal barrier functions in their offspring.
Collapse
|
44
|
Impact of 4-epi-oxytetracycline on the gut microbiota and blood metabolomics of Wistar rats. Sci Rep 2016; 6:23141. [PMID: 26976662 PMCID: PMC4791543 DOI: 10.1038/srep23141] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/29/2016] [Indexed: 01/09/2023] Open
Abstract
The impact of 4-epi-oxytetracycline (4-EOTC), one of the main oxytetracycline (OTC) metabolites, on the gut microbiota and physiological metabolism of Wistar rats was analyzed to explore the dynamic alterations apparent after repeated oral exposure (0.5, 5.0 or 50.0 mg/kg bw) for 15 days as shown by 16S rRNA pyrosequencing and UPLC-Q-TOF/MS analysis. Both principal component analysis and cluster analysis showed consistently altered patterns with distinct differences in the treated groups versus the control groups. 4-EOTC treatment at 5.0 or 50.0 mg/kg increased the relative abundance of the Actinobacteria, specifically Bifidobacteriaceae, and improved the synthesis of lysophosphatidylcholine (LysoPC), as shown by the lipid biomarkers LysoPC(16:0), LysoPC(18:3), LysoPC(20:3), and LysoPC(20:4). The metabolomic analysis of urine samples also identified four other decreased metabolites: diacylglycerol, sphingomyelin, triacylglycerol, and phosphatidylglycerol. Notably, the significant changes observed in these biomarkers demonstrated the ongoing disorder induced by 4-EOTC. Blood and urine analysis revealed that residual 4-EOTC accumulated in the rats, even two weeks after oral 4-EOTC administration, ceased. Thus, through thorough analysis, it can be concluded that the alteration of the gut microbiota and disorders in blood metabolomics are correlated with 4-EOTC treatment.
Collapse
|
45
|
Cerdá B, Pérez M, Pérez-Santiago JD, Tornero-Aguilera JF, González-Soltero R, Larrosa M. Gut Microbiota Modification: Another Piece in the Puzzle of the Benefits of Physical Exercise in Health? Front Physiol 2016; 7:51. [PMID: 26924990 PMCID: PMC4757670 DOI: 10.3389/fphys.2016.00051] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/03/2016] [Indexed: 12/11/2022] Open
Abstract
Regular physical exercise provides many health benefits, protecting against the development of chronic diseases, and improving quality of life. Some of the mechanisms by which exercise provides these effects are the promotion of an anti-inflammatory state, reinforcement of the neuromuscular function, and activation of the hypothalamic-pituitary-adrenal (HPA) axis. Recently, it has been proposed that physical exercise is able to modify gut microbiota, and thus this could be another factor by which exercise promotes well-being, since gut microbiota appears to be closely related to health and disease. The purpose of this paper is to review the recent findings on gut microbiota modification by exercise, proposing several mechanisms by which physical exercise might cause changes in gut microbiota.
Collapse
Affiliation(s)
- Begoña Cerdá
- Research Group on Nutrition, Physical Activity and Health, School of Doctoral Studies and Research, Universidad Europea de Madrid Madrid, Spain
| | - Margarita Pérez
- Research Group on Nutrition, Physical Activity and Health, School of Doctoral Studies and Research, Universidad Europea de Madrid Madrid, Spain
| | - Jennifer D Pérez-Santiago
- Research Group on Nutrition, Physical Activity and Health, School of Doctoral Studies and Research, Universidad Europea de Madrid Madrid, Spain
| | - Jose F Tornero-Aguilera
- Research Group on Nutrition, Physical Activity and Health, School of Doctoral Studies and Research, Universidad Europea de Madrid Madrid, Spain
| | - Rocío González-Soltero
- Research Group on Nutrition, Physical Activity and Health, School of Doctoral Studies and Research, Universidad Europea de Madrid Madrid, Spain
| | - Mar Larrosa
- Research Group on Nutrition, Physical Activity and Health, School of Doctoral Studies and Research, Universidad Europea de Madrid Madrid, Spain
| |
Collapse
|
46
|
SEW2871 protects from experimental colitis through reduced epithelial cell apoptosis and improved barrier function in interleukin-10 gene-deficient mice. Immunol Res 2015; 61:303-11. [PMID: 25588868 DOI: 10.1007/s12026-015-8625-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Loss of intestinal epithelial barrier function including typical tight junction changes and epithelial cell apoptosis plays an important role in Crohn's disease. SEW2871, a selective sphingosine-1-phosphate type-1 receptor agonist, has been proven to be efficient in protecting against colitis in IL-10(-/-) mice in our previous study. Here we performed additional studies to investigate whether treatment with SEW2871 was associated with an improved epithelial barrier function in IL-10(-/-) mice. SEW2871 was administered by gavage at a dose of 20 mg/kg/day for 2 weeks to IL-10(-/-) mice. Severity of colitis, CD4+ T cells in colon lamina propria and proinflammatory cytokine productions were evaluated. Furthermore, intestinal permeability, tight junction (occludin and ZO-1) expressions and distributions, as well as epithelial cell apoptosis, were also assessed. SEW2871 treatment attenuated established colitis associated with decreased CD4+ T cells in colon lamina propria and reduced TNF-α and IFN-γ levels. Moreover, enhanced barrier function, which resulted from ameliorated tight junction (occludin and ZO-1) expressions and suppressed epithelial cell apoptosis, was found to contribute to the therapeutic effects. SEW2871 treatment protects from colitis in IL-10(-/-) mice through reduced epithelial cell apoptosis and improved barrier function. Thus, targeting sphingosine-1-phosphate may represent a new therapeutic approach in Crohn's disease.
Collapse
|
47
|
Sun J, Shen X, Dong J, Zhao J, Zuo L, Wang H, Li Y, Zhu W, Gong J, Li J. Laquinimod ameliorates spontaneous colitis in interleukin-10-gene-deficient mice with improved barrier function. Int Immunopharmacol 2015; 29:423-432. [DOI: 10.1016/j.intimp.2015.10.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 10/16/2015] [Accepted: 10/16/2015] [Indexed: 12/12/2022]
|
48
|
Han SH, Suk KT, Kim DJ, Kim MY, Baik SK, Kim YD, Cheon GJ, Choi DH, Ham YL, Shin DH, Kim EJ. Effects of probiotics (cultured Lactobacillus subtilis/Streptococcus faecium) in the treatment of alcoholic hepatitis: randomized-controlled multicenter study. Eur J Gastroenterol Hepatol 2015; 27:1300-1306. [PMID: 26302024 DOI: 10.1097/meg.0000000000000458] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Probiotics might reduce gut-derived microbial lipopolysaccharide (LPS) by restoring bowel flora in patients with alcoholic hepatitis (AH). We evaluated the therapeutic effects of probiotics in patients with AH. PATIENTS AND METHODS Between September 2010 and April 2012, 117 patients (probiotics 60 and placebo 57) were prospectively randomized to receive the 7 days of cultured Lactobacillus subtilis/Streptococcus faecium (1500 mg/day) or placebo. All patients were hospitalized and were not permitted to consume alcohol for the 7 days of the study. Liver function test, proinflammatory cytokines, LPS, and colony-forming units by stool culture were examined and compared after therapy. RESULTS In both groups, the mean levels of aspartate aminotransferase/alanine aminotransferase, alkaline phosphatase, γ-glutamyl transpeptidase, bilirubin, and prothrombin time were significantly improved after 7 days of abstinence. In the probiotics group (baseline and after), albumin (3.5 ± 0.7 and 3.7 ± 0.6 g/dl, P=0.038) and tumor necrosis factor-α (121 ± 244 and 71 ± 123 pg/ml, P=0.047) showed differences. In addition, the number of colony-forming units of Escherichia coli was significantly reduced (435 ± 287 and 168 ± 210, P=0.002). In the placebo group, the level of LPS (1.7 ± 2.8 and 2.0 ± 2.7 EU/ml) was significantly increased. In the intergroup comparison, significant differences in the levels of tumor necrosis factor-α (P=0.042) and LPS (P=0.028) were observed between the groups. CONCLUSION Immediate abstinence is the most important treatment for patients with AH. In addition, 7 days of oral supplementation with cultured L. subtilis/S. faecium was associated with restoration of bowel flora and improvement of LPS in patients with AH.
Collapse
Affiliation(s)
- Sang Hak Han
- aDepartment of Pathology bDepartment of Internal Medicine cDepartment of Laboratory Medicine, Hallym University College of Medicine dCenter for Efficacy Assessment and Development of Functional Foods and Drugs, Hallym University eDepartment of Internal Medicine, Kangwon University College of Medicine, Chuncheon fDepartment of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju gDepartment of Internal Medicine, Ulsan University College of Medicine, Gangneung hDepartment of Emergency Medical Technology, Daewon University College, Jecheon, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Qiao Y, Qian J, Lu Q, Tian Y, Chen Q, Zhang Y. Protective effects of butyrate on intestinal ischemia–reperfusion injury in rats. J Surg Res 2015; 197:324-30. [DOI: 10.1016/j.jss.2015.04.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/22/2015] [Accepted: 04/09/2015] [Indexed: 12/24/2022]
|
50
|
Wang H, Dong J, Shi P, Liu J, Zuo L, Li Y, Gong J, Gu L, Zhao J, Zhang L, Zhang W, Zhu W, Li N, Li J. Anti-mouse CD52 monoclonal antibody ameliorates intestinal epithelial barrier function in interleukin-10 knockout mice with spontaneous chronic colitis. Immunology 2015; 144:254-62. [PMID: 25087772 DOI: 10.1111/imm.12366] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/30/2014] [Accepted: 07/30/2014] [Indexed: 01/01/2023] Open
Abstract
Intestinal inflammation causes tight junction changes and death of epithelial cells, and plays an important role in the development of Crohn's disease (CD). CD52 monoclonal antibody (CD52 mAb) directly targets the cell surface CD52 and is effective in depleting mature lymphocytes by cytolytic effects in vivo, leading to long-lasting changes in adaptive immunity. The aim of this study was to investigate the therapeutic effect of CD52 mAb on epithelial barrier function in animal models of IBD. Interleukin-10 knockout mice (IL-10(-/-) ) of 16 weeks with established colitis were treated with CD52 mAb once a week for 2 weeks. Severity of colitis, CD4(+) lymphocytes and cytokines in the lamina propria, epithelial expression of tight junction proteins, morphology of tight junctions, tumour necrosis factor-α (TNF-α)/TNF receptor 2 (TNFR2) mRNA expression, myosin light chain kinase (MLCK) expression and activity, as well as epithelial apoptosis in proximal colon were measured at the end of the experiment. CD52 mAb treatment effectively attenuated colitis associated with decreased lamina propria CD4(+) lymphocytes and interferon-γ/IL-17 responses in colonic mucosa in IL-10(-/-) mice. After CD52 mAb treatment, attenuation of colonic permeability, increased epithelial expression and correct localization of tight junction proteins (occludin and zona occludens protein-1), as well as ameliorated tight junction morphology were observed in IL-10(-/-) mice. CD52 mAb treatment also effectively suppressed the epithelial apoptosis, mucosa TNF-α mRNA expression, epithelial expression of long MLCK, TNFR2 and phosphorylation of MLC. Our results indicated that anti-CD52 therapy may inhibit TNF-α/TNFR2-mediated epithelial apoptosis and MLCK-dependent tight junction permeability by depleting activated T cells in the gut mucosa.
Collapse
Affiliation(s)
- Honggang Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|